Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Stroke ; 39(5): 1556-62, 2008 May.
Article in English | MEDLINE | ID: mdl-18323493

ABSTRACT

BACKGROUND AND PURPOSE: Results of our recent pilot clinical trial suggest that the efficacy of thrombolytic therapy in acute ischemic stroke may be enhanced by the coadministration of high-dose albumin. Here, we explored the microvascular hemodynamic effects of this combined therapy in a laboratory model of cortical arteriolar thrombosis. METHODS: We studied the cortical microcirculation of physiologically monitored rats in vivo by two-photon laser-scanning microscopy after plasma-labeling with fluorescein-dextran. We induced focal thrombosis in 30- to 50-microm cortical arterioles by laser irradiation and measured arteriolar flow velocity by repeated line-scanning. At 30 minutes post-thrombosis, we treated animals with the thrombolytic agent, reteplase, which was coadministered with either human albumin, 2 g/kg, or with saline control. RESULTS: Baseline arteriolar flow velocity averaged 3.8+/-0.7 mm/s, was immediately reduced by thrombosis to 22% to 25% of control values, and remained unchanged before treatment. Subthrombolytic doses of reteplase combined with saline led to a median increase in flow velocity to 37% of control distal to the thrombus (P=nonsignificant versus pretreatment). By contrast, reteplase combined with albumin therapy resulted in a prompt, highly significant increase of median flow velocity to 58% of control levels (P=0.013 versus reteplase+saline), which remained significantly higher than the reteplase+saline group at multiple time-points over the subsequent hour. CONCLUSIONS: The beneficial effect of subthrombolytic doses of reteplase on microvascular hemodynamics distal to a cortical arteriolar thrombosis is markedly enhanced by the coadministration of high-dose albumin therapy; these results have important clinical implications for the management of patients with acute ischemic stroke.


Subject(s)
Albumins/pharmacology , Arterioles/drug effects , Brain Ischemia/drug therapy , Cerebrovascular Circulation/drug effects , Intracranial Thrombosis/drug therapy , Thrombolytic Therapy/methods , Albumins/therapeutic use , Animals , Arterioles/pathology , Arterioles/physiopathology , Brain Ischemia/physiopathology , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Cerebral Arteries/radiation effects , Cerebrovascular Circulation/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Intracranial Thrombosis/physiopathology , Lasers/adverse effects , Male , Microscopy, Confocal/methods , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Recovery of Function/drug effects , Recovery of Function/physiology , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
2.
Brain Res ; 1157: 146-55, 2007 Jul 09.
Article in English | MEDLINE | ID: mdl-17543290

ABSTRACT

We characterized acute intracerebral hemorrhage (ICH) in the rat by sequential magnetic resonance imaging (MRI) and correlated MRI findings with neurobehavior and histopathology. In addition, we investigated whether albumin treatment would reduce ICH-induced brain injury. ICH was produced in rats by a double-injection method in which 45 microl of fresh arterial blood was injected into the right striatum. Susceptibility-weighted (SWI) and T2-weighted (T2WI) MRI was carried out on a 4.7T magnet at 0-1 h, 6 h, 24 h, 72 h, and 7 days after ICH. Animals were treated with either 25% human albumin, 1.25 g/kg, or saline vehicle i.v. at 90 min after ICH. Neurological status was evaluated before ICH and after treatment (at 4 h, 24 h, 48 h, 72 h, and 7 days). Brains were then perfusion-fixed, re-imaged on an 11.7T magnet, and studied by histopathology and immunochemistry. MRI revealed a consistent hematoma involving the striatum and overlying corpus callosum, with significant volume changes over time. Lesion volumes computed from T2WI images and by histopathology agreed closely with one another and were highly correlated (p=0.002). SWI lesion volumes were also highly correlated to histological volumes (p<0.001) but overestimated histological hematoma volume by approximately 5-fold. Albumin treatment significantly improved neurological scores compared to saline at 72 h (3.8+/-0.6 vs. 1.5+/-0.7) and 7 days (3.8+/-0.4 vs. 1.3+/-0.5, respectively, p<0.05), but did not affect histological or MRI lesion volumes. Taken together, sequential MRI plus histopathology provides a comprehensive characterization of experimental ICH. Albumin treatment improves neurological deficit after ICH but does not affect MRI or histological hematoma size.


Subject(s)
Albumins/pharmacology , Cerebral Hemorrhage/diagnosis , Cerebral Hemorrhage/drug therapy , Corpus Striatum/drug effects , Corpus Striatum/pathology , Magnetic Resonance Imaging/methods , Albumins/therapeutic use , Animals , Behavior, Animal/physiology , Biomarkers/analysis , Biomarkers/metabolism , Brain Infarction/drug therapy , Brain Infarction/pathology , Brain Infarction/physiopathology , Cerebral Hemorrhage/physiopathology , Corpus Striatum/physiopathology , Disease Models, Animal , Disease Progression , Gliosis/drug therapy , Gliosis/pathology , Gliosis/physiopathology , Immunohistochemistry/methods , Male , Pathology/methods , Rats , Rats, Sprague-Dawley , Staining and Labeling/methods , Time Factors , Treatment Outcome
3.
Acta Neuropathol ; 113(5): 549-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17308915

ABSTRACT

Pathological effects of moderate ischemia (oligemia, hypoperfusion) are relevant in relation to vascular factors in dementia. Chronic bilateral common carotid artery occlusion (BCCAO) in adult Wistar rats induces oligemia and leads to acute changes in gene expression, subacute changes in cortical astrocytes and prolonged changes in white matter tracts, while largely sparing neurons in the forebrain areas. Dilation and remodeling of the basilar artery ensures blood flow to the forebrain. The present study examined the hypoxia-sensitive Purkinje cells in the cerebellum after 6 months of BCCAO using conventional neuropathological analysis, immunohistochemistry and high-precision design-based stereologic methods. Purkinje cells in the vermis region revealed abnormally shaped nuclei. A stereologic analysis showed that the mean total number of Purkinje cells within the vermis was statistically significantly smaller in the BCCAO animals than in the control animals (d = 11.8%; P < 0.0001). BCCAO had no significant effect on the mean volumes of the molecular layer, granule cell layer and white matter in the vermis or the entire cerebellum. Remodeling of the basilar artery indicated that secondary vascular perturbations might be responsible for the effects of BCCAO on the cerebellar Purkinje cells.


Subject(s)
Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Cerebellum/pathology , Purkinje Cells/pathology , Animals , Calbindins , Cell Count/methods , Cell Death/physiology , Fructose-Bisphosphate Aldolase/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Male , Purkinje Cells/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein G/metabolism , Stereotaxic Techniques
4.
Neurosci Lett ; 404(1-2): 170-5, 2006 Aug 14.
Article in English | MEDLINE | ID: mdl-16781056

ABSTRACT

Ischemic preconditioning (IPC) is a phenomenon whereby an organ's adaptive transient resistance to a lethal ischemic insult occurs by preconditioning this organ with a sub-lethal/mild ischemic insult of short duration. Besides IPC, recent studies reported that a short sub-lethal ischemia and reperfusion in various organs can induce ischemic tolerance in another organ as well. This phenomenon is known as remote ischemic preconditioning (RPC). In the present study we tested the hypothesis that tolerance for ischemia can be induced in brain by RPC and IPC in a rat model of asphyxial cardiac arrest (ACA). RPC was induced by tightening the upper two-thirds of both hind limbs using a tourniquet for 15 or 30 min and IPC was induced by tightening bilateral carotid artery ligatures for 2 min. Eight minutes of ACA was induced 48 h after RPC or IPC. After 7 day of resuscitation, brains were extracted and examined for histopathological changes. In CA1 hippocampus, the number of normal neurons was 63% lower in cardiac-arrested rats as compared to the control group. The number of normal neurons in the 15 min RPC, 30 min RPC, and IPC groups was higher than the ACA group by 54, 70, and 67%, respectively. This study demonstrates that RPC and IPC are able to provide neuroprotection in a rat model of ACA. Besides direct application of RPC or IPC paradigms, the exploration of the mechanisms of observed neuroprotection by RPC and IPC may also lead to a possible therapy for CA patients.


Subject(s)
Asphyxia/physiopathology , Brain Ischemia/prevention & control , Heart Arrest/physiopathology , Ischemic Preconditioning , Animals , Brain Ischemia/pathology , Disease Models, Animal , Hippocampus/pathology , Hippocampus/physiopathology , Male , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley
5.
J Neurosci Res ; 82(5): 665-73, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16247804

ABSTRACT

Excitotoxicity is recognized to play a major role in cerebral ischemia-induced cell death. The main goal of the present study was to define whether our model of ischemic preconditioning (IPC) promotes a shift from excitatory to inhibitory neurotransmission during the test ischemia to diminish metabolic demand during the reperfusion phase. We also determined whether gamma-aminobutyric acid (GABA) played a role in IPC-induced neuroprotection. Ten minutes of cerebral ischemia was produced by tightening the carotid ligatures bilaterally following hypotension. Samples of microdialysis perfusate, representing extracellular fluid, were analyzed for amino acid content by HPLC. IPC promoted a robust release of GABA after lethal ischemia compared with control rats. We also observed that the activity of glutamate decarboxylase (the predominant pathway of GABA synthesis in the brain) was higher in the IPC group compared with control and ischemic groups. Because GABAA receptor up-regulation has been shown to occur following IPC, and GABAA receptor activation has been implicated in neuroprotection against ischemic insults, we tested the hypothesis that GABAA or GABAB receptor activation was neuroprotective during ischemia or early reperfusion by using an in vitro model (organotypic hippocampal slice culture). Administration of the GABAB agonist baclofen during test ischemia and for 1 hr of reperfusion provided significant neuroprotection. We concluded that increased GABA release in preconditioned animals after ischemia might be one of the factors responsible for IPC neuroprotection. Specific activation of GABAB receptor contributes significantly to neuroprotection against ischemia in organotypic hippocampal slices.


Subject(s)
Brain Ischemia/metabolism , Cell Death/physiology , Glutamic Acid/biosynthesis , Ischemic Preconditioning , Nerve Degeneration/metabolism , gamma-Aminobutyric Acid/biosynthesis , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Extracellular Fluid/metabolism , GABA Agonists/pharmacology , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurotoxins/metabolism , Organ Culture Techniques , Presynaptic Terminals/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism
6.
Brain Res ; 1052(1): 28-39, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16023090

ABSTRACT

The effects of oligemia (moderate ischemia) on the brain need to be explored because of the potential role of subtle microvascular changes in vascular cognitive impairment and dementia. Chronic bilateral common carotid artery occlusion (BCCAO) in adult rats has been used to study effects of oligemia (hypoperfusion) using neuropathological and neurochemical analysis as well as behavioral tests. In this study, BCCAO was induced for 1 week, or 2, 4, and 6 months. Sensitive immunohistochemistry with marker proteins was used to study reactions of astrocytes (GFAP, nestin), and lectin binding to study microglial cells during BCCAO. Overt neuronal loss was visualized with NeuN antibodies. Astrocytes reacted to changes in the optic tract at all time points, and strong glial reactions also occurred in the target areas of retinal fibers, indicating damage to the retina and optic nerve. Astrocytes indicated a change in the corpus callosum from early to late time points. Diffuse increases in GFAP labeling occurred in parts of the neocortex after 1 week of BCCAO, in the absence of focal changes of neuronal marker proteins. No significant differences emerged in the cortex at longer time points. Nestin labeling was elevated in the optic tract. Reactions of microglia cells were seen in the cortex after 1 week. Measurements of the basilar artery indicated a considerable hypertrophy, indicative of macrovascular compensation in the chronic occlusion model. These results indicate that chronic BCCAO and, by inference, oligemia have a transient effect on the neocortex and a long-lasting effect on white matter structures.


Subject(s)
Arterial Occlusive Diseases/complications , Astrocytes/pathology , Brain Ischemia/pathology , Carotid Artery, Common , Prosencephalon/pathology , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/etiology , Brain Ischemia/metabolism , Carotid Artery, Common/pathology , Cell Count/methods , Chronic Disease , Diagnostic Imaging/methods , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Intermediate Filament Proteins/metabolism , Male , Nerve Tissue Proteins/metabolism , Nestin , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Time Factors
7.
Brain Res ; 1045(1-2): 150-6, 2005 May 31.
Article in English | MEDLINE | ID: mdl-15910773

ABSTRACT

In recent experimental studies, a selective antagonist of endothelin ET(A) receptors, SB 234551, improved neurological and histological outcome in both head trauma and transient focal cerebral ischemia. The present study was conducted to ascertain the degree to which hemodynamic alterations are responsible for this therapeutic effect in a model of permanent middle cerebral artery occlusion (MCAo) in rats. Anesthetized Sprague-Dawley rats were subjected to permanent MCAo by insertion of an intraluminal nylon suture coated with poly-L-lysine. The agent (SB 234551, 30 microg/kg/min = 1.8 mg/kg/h) or vehicle (PBS; 0.6 ml/h) was administered by i.v. infusion beginning 15 min after onset of MCAo and lasting for 23.75 h. Autoradiographic measurement of local cerebral blood flow (lCBF) was performed at 24 h. Physiological data were similar among groups. SB 234551 augmented perfusion by 1.7- to 1.8-fold in both the ischemic hemisphere and in the contralateral (non-ischemic) hemisphere when compared to vehicle-treated ischemic animals. In the ischemic hemisphere, the brain regions significantly benefited were those lying outside the zone of most dense ischemia (i.e., paramedian cortex and thalamus), while in the non-ischemic hemisphere all regions measured showed significant lCBF augmentation. This study demonstrates that SB 234551 therapy results in significant improvement of local cerebral perfusion in the ischemic as well as in the non-ischemic hemispheres after permanent MCAo.


Subject(s)
Brain Ischemia/drug therapy , Cerebral Arteries/drug effects , Cerebral Infarction/drug therapy , Cerebrovascular Circulation/drug effects , Dioxoles/pharmacology , Endothelin A Receptor Antagonists , Pyrazoles/pharmacology , Animals , Brain/blood supply , Brain/drug effects , Brain/physiopathology , Brain Ischemia/physiopathology , Brain Ischemia/prevention & control , Cerebral Arteries/physiology , Cerebral Infarction/physiopathology , Cerebral Infarction/prevention & control , Cerebrovascular Circulation/physiology , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology , Vasodilator Agents/pharmacology
8.
Stroke ; 36(5): 1071-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15790949

ABSTRACT

BACKGROUND AND PURPOSE: Darbepoetin alfa is a novel erythropoiesis-stimulating protein developed for treating anemia. In animal models, exogenous recombinant human erythropoietin has been reported to be beneficial in treating experimental cerebral ischemia. In this study, we determined whether darbepoetin alfa would protect in a rat model of transient focal cerebral ischemia. METHODS: Rats received 2-hour middle cerebral artery suture-occlusion. The drug (darbepoetin alfa, 10 microg/kg) or vehicle was administered intraperitoneally 2 hours after onset of middle cerebral artery occlusion. Animals were allowed to survive for 3 or 14 days. Behavioral tests were performed sequentially. Infarct volumes and brain swelling were determined. RESULTS: Darbepoetin alfa-treated rats showed improved neuroscores relative to vehicle-treated animals beginning within 1 hour of treatment and persisting throughout the 14-day survival period. Darbepoetin alfa significantly reduced corrected total (cortical + subcortical) infarct volume (56.3+/-20.6 and 110.8+/-6.8 mm3, respectively) and total infarct areas at multiple levels compared with vehicle in the 14-day survival group. Brain swelling was not affected by treatment. CONCLUSIONS: Darbepoetin alfa confers behavioral and histological neuroprotection after focal ischemia in rats.


Subject(s)
Brain Ischemia/drug therapy , Erythropoietin/analogs & derivatives , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Behavior, Animal/drug effects , Brain Edema/drug therapy , Brain Edema/pathology , Brain Ischemia/diagnosis , Darbepoetin alfa , Erythropoietin/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use
9.
J Cereb Blood Flow Metab ; 25(6): 730-41, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15716854

ABSTRACT

Protein kinase C (PKC) isozymes have been known to mediate a variety of complex and diverse cellular functions. deltaPKC has been implicated in mediating apoptosis. Using two models of cerebral ischemia, cardiac arrest in rats and oxygen glucose deprivation (OGD) in organotypic hippocampal slices, we tested whether an ischemic insult promoted deltaPKC cleavage during the reperfusion and whether the upstream pathway involved release of cytochrome c and caspase 3 cleavage. We showed that cardiac arrest/OGD significantly enhanced deltaPKC translocation and increased its cleavage at 3 h of reperfusion. Since deltaPKC is one of the substrates for caspase 3, we next determined caspase 3 activation after cardiac arrest and OGD. The maximum decrease in levels of procaspase 3 was observed at 3 h of reperfusion after cardiac arrest and OGD. We also determined cytochrome c release, since it is upstream of caspase 3 activation. Cytochrome c in cytosol increased at 1 h of reperfusion after cardiac arrest/OGD. Inhibition of either deltaPKC/caspase 3 during OGD and early reperfusion resulted in neuroprotection in CA1 region of hippocampus. Our results support the deleterious role of deltaPKC in reperfusion injury. We propose that early cytochrome c release and caspase 3 activation promote deltaPKC translocation/cleavage.


Subject(s)
Brain Ischemia/metabolism , Heart Arrest/metabolism , Nerve Degeneration/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Blood Pressure , Brain Ischemia/pathology , Caspase 3 , Caspases/metabolism , Cell Death/physiology , Cytochromes c/metabolism , Electrocardiography , Glucose/metabolism , Glucose/pharmacology , Heart Arrest/pathology , Hippocampus/enzymology , Hippocampus/pathology , Nerve Degeneration/pathology , Organ Culture Techniques , Oxygen/metabolism , Oxygen/pharmacology , Protein Kinase C-delta , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 313(3): 1090-100, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15716383

ABSTRACT

Stilbazulenyl nitrone (STAZN) is a potent lipophilic second-generation azulenyl nitrone antioxidant, which is highly neuroprotective in rodent models of cerebral ischemia and trauma. This study was conducted to establish whether the neuroprotection induced by STAZN persists with chronic survival and to characterize STAZN's pharmacokinetics. Physiologically regulated rats received a 2-h middle cerebral artery occlusion by intraluminal suture and were treated with either STAZN [four 0.6 mg/kg doses i.p. administered at 2 (i.e., onset of recirculation), 4, 24, and 48 h; n = 16] or dimethyl sulfoxide vehicle (n = 11). They received sequential neurobehavioral examinations followed by quantitative neuropathology at 30 days. STAZN improved neurological deficits compared with vehicle controls, beginning within <2 h of the first dose and persisting throughout a 30-day survival. Large cystic necrotic infarcts were common in vehicle-treated rats but infrequent in STAZN-treated rats, and noninfarcted forebrain tissue was increased on average by 15%. In normal rats administered 5 mg/kg STAZN i.v. in Solutol HS 15/ethanol/saline vehicle, STAZN blood levels exhibited a biexponential decline, with an initial half-life of 28 min and a subsequent slow decay with half-life of approximately 7 h. STAZN tissue levels at 2 to 3 h were, on average, 2.5% of blood levels in forebrain, 56% in myocardium, and 41% in kidney. STAZN was concentrated in liver with initial concentrations averaging 5.2-fold above blood levels and a subsequent linear decline of 40% between 24 and 72 h. These results establish that STAZN confers enduring ischemic neuroprotection, has a long circulating half-life, and penetrates well into brain and other organs-characteristics favoring its potential therapeutic utility.


Subject(s)
Antioxidants/pharmacology , Brain Ischemia/drug therapy , Neuroprotective Agents/pharmacology , Nitrogen Oxides/pharmacology , Animals , Brain Ischemia/pathology , Male , Nitrogen Oxides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Sesquiterpenes
11.
Stroke ; 36(2): 326-31, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15637329

ABSTRACT

BACKGROUND AND PURPOSE: Acute intracerebral hemorrhage (ICH) is a common and severe form of stroke. To date, medical management of ICH has had scant impact on morbidity and mortality. Because albumin therapy is markedly neuroprotective in preclinical models of ischemic stroke, and because ischemic and hemorrhagic stroke share several common injury mechanisms, we hypothesized that albumin therapy might also benefit ICH. METHODS: Acute intracortical hematoma was produced in anesthetized, normothermic rats by the single stereotaxic injection of 50 muL of autologous, nonheparinized whole blood over 5 minutes. Separate animal groups were treated either with 25% human albumin, 1.25 g/kg, or with intravenous saline vehicle at 60 minutes after ICH. Neurobehavior was quantified sequentially over the next 2 to 7 days. Damage to the blood-brain barrier was assessed at 2 days after ICH by fluorometric measurement of Evans blue extravasation in dissected brain regions. RESULTS: High-grade neurological deficits were present in all rats at 50 minutes after ICH (score 10.3+/-0.2, mean+/-SEM [maximal score 12]). Albumin-treated rats showed improved neuroscores relative to saline-treated animals beginning within hours of treatment and persisting throughout the 7-day survival period. At 3 and 7 days, mean total neuroscores of the albumin group were 38% to 43% lower than in saline-treated animals. Perihematomal Evans blue discoloration was readily evident in saline-treated ICH rats but was reduced by albumin treatment. Hemispheric Evans blue content ipsilateral to the hematoma was reduced by 49% by albumin treatment (albumin 93.9+/-13.3 versus saline 184.7+/-33.7 mg/g, P<0.05). Hematoma volume and brain swelling were not affected by albumin treatment. CONCLUSIONS: Prompt albumin therapy improves neurological function and blood-brain barrier integrity after acute intracortical hematoma. These observations have important potential clinical implications.


Subject(s)
Albumins/pharmacology , Blood-Brain Barrier , Cerebral Hemorrhage/diagnosis , Albumins/metabolism , Albumins/therapeutic use , Animals , Brain/pathology , Cerebral Hemorrhage/therapy , Evans Blue/pharmacology , Fluorometry , Hematoma/metabolism , Hematoma/pathology , Humans , Male , Models, Biological , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Sodium Chloride/pharmacology , Time Factors
12.
Stroke ; 36(1): 118-23, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15569878

ABSTRACT

BACKGROUND AND PURPOSE: High-dose human albumin therapy is strongly neuroprotective in models of brain ischemia and trauma and is currently being studied in a pilot-phase clinical stroke trial. Among its actions in ischemia, albumin induces the systemic mobilization of n-3 polyunsaturated fatty acids and may help to replenish polyunsaturated fatty acids lost from neural membranes. METHODS: We complexed 25% human albumin to docosahexaenoic acid (DHA; 22:6n-3) and compared its neuroprotective efficacy with that of native albumin in rats with 2-hour focal ischemia produced by intraluminal suture-occlusion of the middle cerebral artery. RESULTS: In animals treated with DHA-albumin, 0.63 g/kg, the improvement in neurobehavioral scores at 72 hours significantly exceeded that of other treatment groups, and the extent of histological protection (86% reduction in cortical infarction) was highly significant and tended to surpass the degree of cortical protection produced by native albumin at 1.25 g/kg (65%). DHA-albumin 0.63 g/kg, but not native albumin, also significantly reduced subcortical infarction and markedly diminished brain swelling. Lipidomic analysis of DHA-albumin-treated postischemic brains revealed a large accumulation of the neuroprotective DHA metabolite, 10,17S-docosatriene, in the ipsilateral hemisphere. CONCLUSIONS: The high-grade neuroprotection afforded by the DHA-albumin complex at relatively low albumin doses is clinically advantageous in that it might reduce the likelihood of acute intravascular volume overload and congestive heart failure sometimes induced when patients with compromised cardiovascular function are treated with high-dose albumin.


Subject(s)
Brain Ischemia/prevention & control , Docosahexaenoic Acids/therapeutic use , Neuroprotective Agents/therapeutic use , Serum Albumin/therapeutic use , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacokinetics , Male , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reflex , Serum Albumin/pharmacokinetics
13.
Brain Res ; 1024(1-2): 89-96, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15451369

ABSTRACT

Cardiac arrest (CA) patients exhibit learning and memory disabilities. These deficits suggest that synaptic dysfunction may underlie such disabilities. The hypothesis of the present study was that synaptic dysfunction occurs following CA and that this precedes cell death. To test this hypothesis, we used histopathological and electrophysiological markers in the hippocampus of rats subjected to CA. Evoked potentials (EP) were determined in the CA1 region of hippocampal slices harvested from animals subjected to CA or sham-operated rats by stimulating the Schaffer collaterals and recording in the CA1 pyramidal region. EP amplitudes were significantly attenuated by approximately 60% in hippocampal slices harvested from animals subjected to CA. Hippocampal slices harvested from sham rats exhibited normal long-term potentiation (LTP). In contrast, hippocampal slices harvested 24 h after CA exhibited no LTP response, even when no histopathological abnormalities were observed. These data suggest that synaptic dysfunction occurs before and without overt histopathology. We suggest that the synaptic dysfunction precedes and may be an early marker for delayed neuronal cell death in the hippocampus after CA.


Subject(s)
Action Potentials/physiology , Heart Arrest/physiopathology , Hippocampus/physiopathology , Synapses/physiology , Animals , Blood Pressure/physiology , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley
14.
Brain Res ; 1008(2): 278-83, 2004 May 22.
Article in English | MEDLINE | ID: mdl-15145766

ABSTRACT

The combination of low-dose ethanol and caffeine (caffeinol) protects cortical areas of the brain from damage produced by distal focal ischemia in rats. There are no data, however, as to whether caffeinol influences injury in subcortical brain regions. Rats were anesthetized with halothane and subjected to 2 h of MCAo by poly-l-lysine-coated intraluminal suture. Caffeinol [a combination of ethanol, 0.33 g/kg, and caffeine, 10 mg/kg (n=5)] or vehicle (0.9% NaCl; n=7) was administered by i.v. infusion over a 2.5-h period beginning 15 min after reperfusion. Neurological status was evaluated daily, and histopathology was quantified at 3 days. Caffeinol therapy significantly improved the neurological score, reduced the total infarct volume (by 52%) and cortical infarct areas at multiple coronal levels, but subcortical infarction and brain swelling were not affected.


Subject(s)
Caffeine/pharmacology , Cerebral Cortex/pathology , Ethanol/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Caffeine/administration & dosage , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Infarction, Middle Cerebral Artery/pathology , Infusions, Intravenous , Ischemic Attack, Transient/pathology , Male , Movement/physiology , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
15.
Brain Res ; 1001(1-2): 133-42, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14972662

ABSTRACT

The 19 kD interacting protein 3, Nip3/BNIP3, is a pro-apoptotic member of the Bcl-2 family induced during hypoxia via the hypoxia-inducible factor (HIF) 1. BNIP3 has been linked to both apoptotic and necrotic cell death involving mitochondrial permeability transition. Since apoptotic and necrotic mechanisms may occur in brain ischemia, immunohistochemical changes of BNIP3 were studied at 1, 2, 3 and 7 days after transient global brain ischemia (12.5 min) in ventilated normothermic rats. In control brains, BNIP3-like immunoreactivity was moderately strong in neuronal processes or cytoplasm and absent in the nucleus. In the ischemia-vulnerable CA1 neurons, BNIP3-positive granules were seen in the nucleus at 1 and 2 days, and these neurons were damaged at 3 and 7 days. The resistant CA3 neurons showed nuclear BNIP3 labeling by 1 day and then returned to the normal state. BNIP3-positive granules did not overlap with the nucleolus. Constitutively expressed BNIP3 may participate in apoptotic and necrotic processes after brain ischemia. Nuclear location of BNIP3 after brain ischemia indicates a novel role for the regulation of cell survival in neurons or a general disturbance of the nuclear envelope.


Subject(s)
Brain Ischemia/metabolism , Cell Nucleus/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Western/methods , Cell Count/methods , Computational Biology/methods , Databases, Protein , Hippocampus/pathology , Humans , Immunohistochemistry/methods , Male , Mice , Microscopy, Confocal/methods , Microtubule-Associated Proteins/metabolism , Rats , Rats, Wistar
16.
Brain Res ; 998(2): 243-6, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14751596

ABSTRACT

Human albumin therapy within the first 4 h is highly neuroprotective in focal ischemia, but it is unknown whether delayed albumin therapy is deleterious. Rats received 2 h middle cerebral artery suture-occlusion. Human albumin (25%, 2.5 mg/kg; n=12) or vehicle (0.9% saline, 5 ml/kg; n=9) were administered at 19 h. Neurological status was evaluated daily, and histopathology and brain swelling were quantified at 3 days. Delayed albumin treatment, while ineffective, failed to show adverse effects.


Subject(s)
Albumins/therapeutic use , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Neuroprotective Agents/therapeutic use , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Brain Edema/drug therapy , Humans , Male , Rats , Rats, Sprague-Dawley , Time Factors
17.
Article in English | MEDLINE | ID: mdl-15799556

ABSTRACT

BACKGROUND: Vascular endothelial growth factor and mitochondrial abnormalities have been described in ALS and its animal models. We have reported that hyperbaric oxygen (HBO) treatment delayed the onset of weakness in the wobbler mouse. OBJECTIVE: To perform a Phase I safety study of HBO in patients with ALS. METHODS: Five patients with ALS were treated for 60min with 100% oxygen at 2 atmospheres pressure daily for five days a week for four weeks. The patients reported any deterioration in their condition after each treatment, and their neurological condition was measured serially during the four weeks of the treatment, and for four further weeks. RESULTS: Four patients reported decreased fatigue, while one patient dropped out at three weeks because of increased fatigue. Maximum isometric voluntary contraction (MVIC) of all muscle groups except right hand grip improved significantly by up to 97%. Most improvement occurred during the four weeks after treatment. It is possible that the improvement in muscle strength was a placebo or a learning effect, though no such effects have been detected in prior therapeutic trials in ALS using MVIC. No change was detected in other measures of neuromuscular function. CONCLUSIONS: A longer duration, placebo controlled trial in a larger number of patients is needed to determine the safety and efficacy of HBO. Until that is completed, it is not recommended that ALS patients should be treated with HBO.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/therapy , Hyperbaric Oxygenation/adverse effects , Hyperbaric Oxygenation/methods , Aged , Female , Humans , Linear Models , Male , Middle Aged
18.
Ann Neurol ; 54(3): 330-42, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12953265

ABSTRACT

Azulenyl nitrones are novel chain-breaking antioxidants with low oxidation potentials and high lipophilicity-properties favoring their efficacy as neuroprotectants. We tested the second-generation azulenyl nitrone, stilbazunenlyl nitrone (STAZN), in focal ischemic stroke. Physiologically monitored rats received 2 hours of middle cerebral artery occlusion by intraluminal suture, resulting in substantial cortical and striatal infarcation. Neurobehavior was quantified on a standard battery, and brains were perfusion-fixed for quantitative histopathology at 3 days. In 3 independent series, rats were treated at either 2h + 4h, or 2h + 4h + 24h + 48h, after onset of ischemia; vehicle-treated rats received dimethylsulfoxide or saline. All animals (n = 52) developed high-grade neurological deficits (score 11 of 12) during ischemia, which improved, in STAZN-treated rats, within 1-1.5 h of the initial dose and fell to a median score of 3 at 72 h, compared to 8 in vehicle rats. STAZN treatment reduced mean cortical infarct volume by 64-97%, and total infarct volume by 42-72%. In over one-half of STAZN-treated animals, cortical infarction was virtually abolished. Regression analysis predicted that STAZN would confer approximately 50% cortical neuroprotection even in the most severely affected cases. The potency of STAZN was orders-of-magnitude greater than other nitrones such as NXY-059. These results suggest that STAZN has great promise for ischemic stroke.


Subject(s)
Brain/drug effects , Brain/pathology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Nitrogen Oxides/therapeutic use , Animals , Antioxidants/therapeutic use , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/physiopathology , Brain Edema/drug therapy , Dimethyl Sulfoxide/pharmacology , Disease Models, Animal , Free Radical Scavengers/pharmacology , Image Processing, Computer-Assisted , Infarction, Middle Cerebral Artery/pathology , Male , Neuroprotective Agents/blood , Nitrogen Oxides/blood , Rats , Rats, Sprague-Dawley , Sesquiterpenes
19.
Stroke ; 34(9): 2221-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12920264

ABSTRACT

BACKGROUND AND PURPOSE: A major limitation of intracerebral hemorrhage (ICH) research is the lack of reproducible animal models. The present study was conducted to validate in the mouse the double-injection method of ICH initially developed in the rat. We investigated the effect of intrastriatal injection of blood or cerebrospinal fluid (CSF) on cerebral blood flow (CBF), neurological score, hematoma volume, and brain swelling. METHODS: Male C57BL/6 mice were anesthetized with halothane/nitrous oxide delivered by face mask. Rectal and cranial temperatures were regulated at 37 degrees C to 37.5 degrees C. Mice were placed in a stereotactic frame, and a 30-gauge stainless steel cannula was introduced through a burr hole into the left striatum. Each mouse received a 5-microL injection of either whole blood or CSF (over 3 minutes), followed 7 minutes later by 10 microL injected over 5 minutes. The injection cannula was slowly withdrawn 10 minutes after the second injection. Control mice had only cannula insertion. CBF was studied by laser Doppler perfusion imaging. Neurological status was evaluated on days 1 and 2. After 2 days, hematoma volume and brain swelling were calculated. RESULTS: Physiological values were stable. Mice with ICH but not those with CSF or cannula alone had a marked, persistent neurological deficit and a highly reproducible hematoma, whose mean+/-SEM volume was 2.0+/-0.2 mm3 compared with a lesion size of 0.2+/-0.1 mm3 in mice with CSF. Residual swelling of the ipsilateral hemisphere at 48 hours was 5.7% in the hematoma and 1.5% in the CSF groups. Relative CBF in the neocortex ipsilateral to the injection site declined by approximately 45% to 60% during the first 20 minutes after cannula insertion/injection in all groups but began to renormalize at approximately 25 to 30 minutes in the CSF and cannula-only groups; in the hematoma group, cortical hypoperfusion of approximately 35% to 50% persisted during the 90-minute measurement period. CONCLUSIONS: The present ICH model in mice produces a consistent neurological deficit, hypoperfusion, hematoma volume, and brain swelling. This model closely mimics human hypertensive basal ganglionic ICH and should be useful for the evaluation of pharmaceutical therapies. Laser Doppler perfusion imaging is a useful new technique to quantify relative CBF changes and can be used for studies of dynamic changes of CBF in this in vivo model of ICH in mice.


Subject(s)
Behavior, Animal , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Disease Models, Animal , Hemodynamics , Animals , Basal Ganglia Hemorrhage/pathology , Basal Ganglia Hemorrhage/physiopathology , Blood , Blood Flow Velocity , Cerebrospinal Fluid , Cerebrovascular Circulation , Corpus Striatum/blood supply , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Disease Progression , Injections , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Stereotaxic Techniques
20.
Stroke ; 34(3): 758-63, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12624304

ABSTRACT

BACKGROUND AND PURPOSE: SolCD39 is a soluble form of recombinant human ecto-ATP/ADPase (NTPDase1) and represents a new class of antithrombotic agents. SolCD39 blocks and reverses platelet activation, preventing recruitment of additional platelets into a growing thrombus. The purpose of this study was to examine the effect of solCD39 on neurological deficit, infarct size, and extent of edema after transient middle cerebral artery occlusion (MCAO) in rats. METHODS: Physiologically controlled Sprague-Dawley rats underwent 2-hour MCAO by retrograde insertion of an intraluminal suture coated with poly-l-lysine. The agent (solCD39) was administered intravenously before MCAO or at 1-hour or 3-hour recirculation. Other groups received vehicle (Tris-buffered saline) or human albumin (as a "positive" neuroprotective control; 25%, 0.5% of body weight) at 1-hour recirculation. Neurological status was evaluated during occlusion (at 60 minutes) and daily for 3 days after MCAO. Brains were perfusion-fixed at 72 hours, and infarct volumes and brain swelling were determined. RESULTS: Pretreatment with solCD39 significantly improved the neurological score at 72 hours compared with the vehicle group (4.4+/-0.6 versus 7.6+/-0.6, respectively; P=0.008). Cortical infarct areas were significantly reduced at multiple levels by pretreatment with solCD39. Total striatal infarct area was also significantly reduced compared with vehicle by both solCD39 pretreatment (48% mean reduction) and solCD39 treatment at 3-hour recirculation (51% mean reduction). Treatment with SolCD39 significantly reduced total infarct volume (corrected for brain swelling) by an average of 71% to 72% when administered either before ischemia or at 3 hours of recirculation compared with vehicle. Treatment with albumin significantly reduced neurological score and total, cortical, and subcortical infarction at multiple levels, as expected. CONCLUSIONS: Treatment with SolCD39, administered either before or at 3 hours after MCAO, improves neurological score and reduces infarct size compared with vehicle. A pharmacological agent of this type appears to have potential for the treatment of focal ischemic stroke.


Subject(s)
Adenosine Triphosphatases/pharmacology , Antigens, CD/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Animals , Apyrase , Behavior, Animal/drug effects , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Edema/etiology , Brain Edema/pathology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Ischemic Attack, Transient/etiology , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Serum Albumin/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...